外科理论与实践 ›› 2025, Vol. 30 ›› Issue (04): 351-357.doi: 10.16139/j.1007-9610.2025.04.10
李雅琪, 莫少波 综述, 彭俊杰 审校
收稿日期:2024-04-03
出版日期:2025-07-25
发布日期:2025-10-23
基金资助:LI Yaqi, MO Shaobo, PENG Junjie
Received:2024-04-03
Online:2025-07-25
Published:2025-10-23
摘要:
循环肿瘤DNA(ctDNA)作为一种非侵入性生物标志物,能敏感地识别微小残留病灶(MRD),可提早发现复发转移,为结肠直肠癌肝转移(CRLM)病人的预后预测和疗效评估提供了新方法,有助于指导个性化治疗方案的制定。本文归纳了ctDNA检测MRD在CRLM中的应用进展,并对其未来发展方向加以展望。
中图分类号:
李雅琪, 莫少波, 彭俊杰. 循环肿瘤DNA检测微小残留病灶在结肠直肠癌肝转移中的应用进展[J]. 外科理论与实践, 2025, 30(04): 351-357.
LI Yaqi, MO Shaobo, PENG Junjie. Progression in circulating tumor DNA detection for minimal residual disease in patients with colorectal cancer liver metastasis[J]. Journal of Surgery Concepts & Practice, 2025, 30(04): 351-357.
表1
ctDNA检测策略的主要技术特点及应用范围
| Technical route | Tumor-informed assay | Tumor-agnostic assay |
|---|---|---|
| Panel type | Whole exome or whole genome sequencing + personalized panel /multiple PCR | Non-personalized, fixed panel |
| Tumor tissue | Required | Not required |
| Detection genes | 16-40 genes | 200-500 genes |
| Detection level | Genomic | Genomic and epigenomic |
| Initial detection cycle | 4-6 weeks | 7-10 days |
| Subsequent detection cycle | 7-10 days | 7-10 days |
| Limit of detection | 0.02%-0.005% | 0.1%-0.05% |
| Cancer type applicability | Not affected by tumor type, widely applicable to various cancers | Can simultaneously detect multiple cancer types or highly heterogeneous tumors |
| Main advantages | High sensitivity, high accuracy | Do not require tumor tissue, lower cost |
| Main disadvantages | Requires tumor tissue sample, longer customization time, higher cost | High false-positive rate |
| [1] | SIEGEL R L, GIAQUINTO A N, JEMAL A. Cancer statistics,2024[J]. CA Cancer J Clin, 2024, 74(1):12-49. |
| [2] | HAN B, ZHENG R, ZENG H, et al. Cancer incidence and mortality in China, 2022[J]. J Natl Cancer Cent, 2024, 4(1):47-53. |
| [3] |
ABDALLA E K, VAUTHEY J N, ELLIS L M, et al. Recurrence and outcomes following hepatic resection, radiofrequency ablation, and combined resection/ablation for colorectal liver metastases[J]. Ann Surg, 2004, 239(6):818-825.
doi: 10.1097/01.sla.0000128305.90650.71 pmid: 15166961 |
| [4] | DEVAUD N, KANJI Z S, DHANI N, et al. Liver resection after chemotherapy and tumour downsizing in patients with initially unresectable colorectal cancer liver metastases[J]. HPB (Oxford), 2014, 16(5):475-480. |
| [5] | LEAL J N, BRESSAN A K, VACHHARAJANI N, et al. Time-to-surgery and survival outcomes in resectable colorectal liver metastases: a multi-institutional evaluation[J]. J Am Coll Surg, 2016, 222(5):766-779. |
| [6] |
MARGONIS G A, SERGENTANIS T N, NTANASIS-STATHOPOULOS I, et al. Impact of surgical margin width on recurrence and overall survival following R0 hepatic resection of colorectal metastases: a systematic review and meta-analysis[J]. Ann Surg, 2018, 267(6):1047-1055.
doi: 10.1097/SLA.0000000000002552 pmid: 29189379 |
| [7] | MAHURON K M, FONG Y. Applications of liquid bio-psy for surgical patients with cancer: a review[J]. JAMA Surg, 2024, 159(1):96-103. |
| [8] | LOFT M, TO Y H, GIBBS P, et al. Clinical application of circulating tumour DNA in colorectal cancer[J]. Lancet Gastroenterol Hepatol, 2023, 8(9):837-852. |
| [9] | MALLA M, LOREE J M, KASI P M, et al. Using circula-ting tumor DNA in colorectal cancer: current and evolving practices[J]. J Clin Oncol, 2022, 40(24):2846-2857. |
| [10] |
SATO R, SEMBA T, SAYA H, et al. Concise review: stem cells and epithelial-mesenchymal transition in cancer: biological implications and therapeutic targets[J]. Stem Cells, 2016, 34(8):1997-2007.
doi: 10.1002/stem.2406 pmid: 27251010 |
| [11] | RING A, NGUYEN-STRÄULI B D, WICKI A, et al. Bio-logy, vulnerabilities and clinical applications of circulating tumour cells[J]. Nat Rev Cancer, 2023, 23(2):95-111. |
| [12] |
MASHOURI L, YOUSEFI H, AREF A R, et al. Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance[J]. Mol Cancer, 2019, 18(1):75.
doi: 10.1186/s12943-019-0991-5 pmid: 30940145 |
| [13] | ZHANG L, YU D. Exosomes in cancer development, metastasis, and immunity[J]. Biochim Biophys Acta Rev Cancer, 2019, 1871(2):455-468. |
| [14] |
WU J, HU S, ZHANG L, et al. Tumor circulome in the liquid biopsies for cancer diagnosis and prognosis[J]. Theranostics, 2020, 10(10):4544-4556.
doi: 10.7150/thno.40532 pmid: 32292514 |
| [15] |
HENRIKSEN T V, DRUE S O, FRYDENDAHL A, et al. Error characterization and statistical modeling improves circulating tumor DNA detection by droplet digital PCR[J]. Clin Chem, 2022, 68(5):657-667.
doi: 10.1093/clinchem/hvab274 pmid: 35030248 |
| [16] |
O'LEARY B, HREBIEN S, BEANEY M, et al. Comparison of BEAMing and droplet digital PCR for circulating tumor DNA analysis[J]. Clin Chem, 2019, 65(11):1405-1413.
doi: 10.1373/clinchem.2019.305805 pmid: 31551314 |
| [17] | NIKANJAM M, KATO S, KURZROCK R. Liquid biopsy: current technology and clinical applications[J]. J Hematol Oncol, 2022, 15(1):131. |
| [18] | NAJAFI S, MAJIDPOOR J, MORTEZAEE K. Liquid bio-psy in colorectal cancer[J]. Clin Chim Acta, 2024,553:117674. |
| [19] | TORRESAN S, DE SCORDILLI M, BORTOLOT M, et al. Liquid biopsy in colorectal cancer: onward and upward[J]. Crit Rev Oncol Hematol, 2024,194:104242. |
| [20] |
LUO H, WEI W, YE Z, et al. Liquid biopsy of methylation biomarkers in cell-free DNA[J]. Trends Mol Med, 2021, 27(5):482-500.
doi: 10.1016/j.molmed.2020.12.011 pmid: 33500194 |
| [21] | NASSAR F J, MSHEIK Z S, NASR R R, et al. Methy-lated circulating tumor DNA as a biomarker for colorectal cancer diagnosis,prognosis, and prediction[J]. Clin Epigenetics, 2021, 13(1):111. |
| [22] | JIN S, ZHU D, SHAO F, et al. Efficient detection and post-surgical monitoring of colon cancer with a multi-marker DNA methylation liquid biopsy[J]. Proc Natl Acad Sci U S A, 2021, 118(5):e2017421118. |
| [23] | LUO H, ZHAO Q, WEI W, et al. Circulating tumor DNA methylation profiles enable early diagnosis, prognosis prediction, and screening for colorectal cancer[J]. Sci Transl Med, 2020, 12(524):eaax7533. |
| [24] | MO S, DAI W, WANG H, et al. Early detection and prognosis prediction for colorectal cancer by circulating tumour DNA methylation haplotypes: a multicentre cohort study[J]. E Clin Med, 2022,55:101717. |
| [25] | MO S, YE L, WANG D, et al. Early detection of molecular residual disease and risk stratification for stage Ⅰ to Ⅲ colorectal cancer via circulating tumor DNA methylation[J]. JAMA Oncol, 2023, 9(6):770-778. |
| [26] |
DAWOOD Z S, ALAIMO L, LIMA H A, et al. Circulating tumor DNA, imaging, and carcinoembryonic antigen: comparison of surveillance strategies among patients who underwent resection of colorectal cancer-a systematic review and meta-analysis[J]. Ann Surg Oncol, 2023, 30(1):259-274.
doi: 10.1245/s10434-022-12641-7 pmid: 36219278 |
| [27] |
KOTANI D, OKI E, NAKAMURA Y, et al. Molecular residual disease and efficacy of adjuvant chemotherapy in patients with colorectal cancer[J]. Nat Med, 2023, 29(1):127-134.
doi: 10.1038/s41591-022-02115-4 pmid: 36646802 |
| [28] | REINERT T, HENRIKSEN T V, CHRISTENSEN E, et al. Analysis of plasma cell-free DNA by ultradeep sequencing in patients with stages Ⅰ to Ⅲ colorectal cancer[J]. JAMA Oncol, 2019, 5(8):1124-1131. |
| [29] | TIE J, WANG Y, TOMASETTI C, et al. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage Ⅱ colon cancer[J]. Sci Transl Med, 2016, 8(346):346ra392. |
| [30] | CHEN G, PENG J, XIAO Q, et al. Postoperative circulating tumor DNA as markers of recurrence risk in stages Ⅱ to Ⅲ colorectal cancer[J]. J Hematol Oncol, 2021, 14(1):80. |
| [31] | LI Y, MO S, ZHANG L, et al. Postoperative circulating tumor DNA combined with consensus molecular subtypes can better predict outcomes in stage Ⅲ colon cancers: a prospective cohort study[J]. Eur J Cancer, 2022,169:198-209. |
| [32] |
TIE J, COHEN J D, WANG Y, et al. Serial circulating tumour DNA analysis during multimodality treatment of locally advanced rectal cancer: a prospective biomarker study[J]. Gut, 2019, 68(4):663-671.
doi: 10.1136/gutjnl-2017-315852 pmid: 29420226 |
| [33] | TIE J, COHEN J D, WANG Y, et al. Circulating tumor DNA analyses as markers of recurrence risk and benefit of adjuvant therapy for stage Ⅲ colon cancer[J]. JAMA Oncol, 2019, 5(12):1710-1717. |
| [34] |
NAKAMURA Y, WATANABE J, AKAZAWA N, et al. ctDNA-based molecular residual disease and survival in resectable colorectal cancer[J]. Nat Med, 2024, 30(11):3272-3283.
doi: 10.1038/s41591-024-03254-6 pmid: 39284954 |
| [35] | TIE J, COHEN J D, LAHOUEL K, et al. Circulating tumor DNA analysis guiding adjuvant therapy in stage Ⅱ colon cancer[J]. N Engl J Med, 2022, 386(24):2261-2272. |
| [36] | TIE J, WANG Y, COHEN J, et al. Circulating tumor DNA dynamics and recurrence risk in patients undergoing curative intent resection of colorectal cancer liver metastases: a prospective cohort study[J]. PLoS Med, 2021, 18(5):e1003620. |
| [37] | WANG D S, YANG H, LIU X Y, et al. Dynamic monitoring of circulating tumor DNA to predict prognosis and efficacy of adjuvant chemotherapy after resection of colorectal liver metastases[J]. Theranostics, 2021, 11(14):7018-7028. |
| [38] |
LI Y, XU J, HU X, et al. Personalized circulating tumor DNA monitoring improves recurrence surveillance and management after curative resection of colorectal liver metastases: a prospective cohort study[J]. Int J Surg, 2024, 110(5):2776-2787.
doi: 10.1097/JS9.0000000000001236 pmid: 38445460 |
| [39] | COHEN S A, LIU M C, ALESHIN A. Practical recommendations for using ctDNA in clinical decision making[J]. Nature, 2023, 619(7969):259-268. |
| [40] | BOLHUIS K, VAN 'T ERVE I, MIJNALS C, et al. Postoperative circulating tumour DNA is associated with pathologic response and recurrence-free survival after resection of colorectal cancer liver metastases[J]. EBioMedicine, 2021,70:103498. |
| [41] | ØGAARD N, REINERT T, HENRIKSEN T V, et al. Tumour-agnostic circulating tumour DNA analysis for improved recurrence surveillance after resection of colorectal liver metastases: a prospective cohort study[J]. Eur J Cancer, 2022,163:163-176. |
| [42] |
REINERT T, PETERSEN L M S, HENRIKSEN T V, et al. Circulating tumor DNA for prognosis assessment and postoperative management after curative-intent resection of colorectal liver metastases[J]. Int J Cancer, 2022, 150(9):1537-1548.
doi: 10.1002/ijc.33924 pmid: 34994972 |
| [43] |
KATAOKA K, MORI K, NAKAMURA Y, et al. Survival benefit of adjuvant chemotherapy based on molecular residual disease detection in resected colorectal liver metastases: subgroup analysis from CIRCULATE-Japan GALAXY[J]. Ann Oncol, 2024, 35(11):1015-1025.
doi: 10.1016/j.annonc.2024.08.2240 pmid: 39293512 |
| [44] | BANDO H, KAGAWA Y, KATO T, et al. A multicentre, prospective study of plasma circulating tumour DNA test for detecting RAS mutation in patients with metastatic colorectal cancer[J]. Br J Cancer, 2019, 120(10):982-986. |
| [45] | KAGAWA Y, ELEZ E, GARCÍA-FONCILLAS J, et al. Combined analysis of concordance between liquid and tumor tissue biopsies for RAS mutations in colorectal cancer with a single metastasis site: the METABEAM study[J]. Clin Cancer Res, 2021, 27(9):2515-2522. |
| [46] |
SAIURA A, YAMAMOTO J, KOGA R, et al. Favorable outcome after repeat resection for colorectal liver metastases[J]. Ann Surg Oncol, 2014, 21(13):4293-4299.
doi: 10.1245/s10434-014-3863-7 pmid: 24962942 |
| [47] |
SALAH S, ARDISSONE F, GONZALEZ M, et al. Pulmonary metastasectomy in colorectal cancer patients with previously resected liver metastasis: pooled analysis[J]. Ann Surg Oncol, 2015, 22(6):1844-1850.
doi: 10.1245/s10434-014-4173-9 pmid: 25326396 |
| [48] | OBA M, HASEGAWA K, SHINDOH J, et al. Survival benefit of repeat resection of successive recurrences after the initial hepatic resection for colorectal liver metastases[J]. Surgery, 2016, 159(2):632-640. |
| [49] | LIU W, LIU J M, WANG K, et al. Recurrent colorectal liver metastasis patients could benefit from repeat hepatic resection[J]. BMC Surg, 2021, 21(1):327. |
| [1] | 赵一鸣, 吴棕, 王鲁. 肝脏微环境细胞对结肠直肠癌肝转移的作用[J]. 外科理论与实践, 2024, 29(02): 126-131. |
| [2] | 潘睿忻 综述, 陈小松, 沈坤炜 审校. 循环肿瘤DNA和循环肿瘤细胞检测乳腺癌微小残留病灶的研究[J]. 外科理论与实践, 2022, 27(05): 463-467. |
| [3] | 包全, 邢宝才. 复杂双叶多发性结肠直肠癌肝转移外科治疗策略[J]. 外科理论与实践, 2022, 27(02): 128-130. |
| [4] | 张欢, 张阳. 进展期胃癌化疗疗效的影像学评估[J]. 外科理论与实践, 2019, 24(01): 6-9. |
| [5] | 金泽宇, 陈小松. 循环肿瘤DNA在乳腺癌诊治中应用的研究[J]. 外科理论与实践, 2018, 23(05): 473-476. |
| [6] | 顾圣佳, 曹琪琪, 严福华, 杨文洁. 双能CT在非小细胞肺癌非手术治疗疗效评估中的应用价值[J]. 诊断学理论与实践, 2018, 17(05): 526-532. |
| [7] | 黄新韵, 胡佳佳, 李彪. 18F-FDG PET/CT显像在淋巴瘤疗效评估中的应用进展[J]. 诊断学理论与实践, 2017, 16(04): 437-441. |
| [8] | 陈冰, 眭竫旎. 急性髓系白血病微小残留病监测方式的展望[J]. 诊断学理论与实践, 2017, 16(01): 17-26. |
| [9] | 张佼佼, 糜坚青,. 微小残留病灶与慢性淋巴细胞白血病预后的相关性研究[J]. 内科理论与实践, 2016, 11(06): 391-396. |
| [10] | 李彪, 张敏,. ~(18)F-FDGPET及PET/CT在淋巴瘤疗效评估中的应用[J]. 诊断学理论与实践, 2012, 11(02): 108-110. |
| [11] | 余栋, 赵维莅,. 慢性淋巴细胞性白血病诊断和治疗进展[J]. 内科理论与实践, 2009, 4(04): 341-345. |
| [12] | 田勇,穆雄铮,柴岗,曹德君. 口内入路下颌角弧形截骨术矫正重度下颌角肥大[J]. 组织工程与重建外科杂志, 2007, 3(3): 147-148. |
| [13] | 曾民德,王泰龄,王宝恩. 肝纤维化诊断和疗效评估共识[J]. 诊断学理论与实践, 2002, 1(03): 67-68. |
| 阅读次数 | ||||||
|
全文 |
|
|||||
|
摘要 |
|
|||||